Chapter 2 Infections in Hematopoietic Stem Cell Transplant Recipients

Chapter 2 Infections in Hematopoietic Stem Cell Transplant Recipients Georg Maschmeyer and Per Ljungman Abstract  The risk of infection among alloge...
3 downloads 0 Views 1MB Size
Chapter 2

Infections in Hematopoietic Stem Cell Transplant Recipients Georg Maschmeyer and Per Ljungman

Abstract  The risk of infection among allogeneic hematopoietic­ stem cell transplant (aHSCT) recipients is determined by patient age, underlying disease, the complications that occurred during preceding treatment regimens, the selected transplantation modality, and the severity of graft-versus-host disease. Immunological reconstitution after hematopoietic recovery has an impact on the type of posttransplant infectious complications, and infectionrelated mortality is significantly higher postengraftment than during the short posttransplant neutropenia. As different pathogenetic and epidemiological backgrounds of infections occur following aHSCT, three consecutive time periods posttransplant are separately described: the early posttransplant period (preengraftment, comprising 3 weeks), the intermediate posttransplant period (3 weeks to 3 months), and the late posttransplant period (later than day + 90). Keywords  Allogeneic • Hematopoietic stem cell transplant • Early infection • CMV • Late infections • Graft-versushost disease

Introduction Fever and Infection After Allogeneic Hematopoietic Stem Cell Transplant The risk of infection among allogeneic hematopoietic stem cell transplant (aHSCT) recipients is determined by patient age, underlying disease, the complications that occurred

G. Maschmeyer (*) Department of Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Charlottenstrasse 72, 14467 Potsdam, Germany e-mail: [email protected]

during­ preceding treatment regimens, the selected transplantation­ modality, and the severity of graft-versushost disease (GvHD) [1, 2]. In comparison with patients undergoing high-dose chemotherapy and autologous stem cell transplantation, recipients of aHSCT are at a much higher risk of infection also after hematopoietic reconstitution, due to delayed recovery of T-cell and B-cell functions. Immunological reconstitution after hematopoietic recovery has an impact on the type of posttransplant infectious complications [3, 4], and infection-related mortality is significantly higher postengraftment than during the short posttransplant neutropenia. After nonmyeloablative conditioning, there is a lower risk of severe and fatal infections in the early posttransplant period [5–9]. Because of different pathogenetic and epidemiological backgrounds of infections, three consecutive time periods posttransplant are separately described: the early posttransplant period (preengraftment, comprising 3 weeks), the intermediate posttransplant period (3 weeks to 3 months), and the late posttransplant period (later than day + 90) (Fig. 2.1).

Early Posttransplant Period (Preengraftment; Earlier than Day +21) Epidemiology of Infections During Neutropenia Posttransplant Almost all patients receiving myeloablative conditioning regimens develop fever during neutropenia, and most of these febrile episodes are due to infections. The risk of severe bacterial or fungal infection in the early posttransplant period is markedly reduced when nonmyeloablative conditioning has been used. Clinical signs of infection apart from fever may be absent or discrete, and an infectious focus frequently will not be identified by clinical examination, microbiological, or imaging techniques. The differential diagnosis of noninfectious causes of fever, such as transfusion reactions,

A. Safdar (ed.), Principles and Practice of Cancer Infectious Diseases, Current Clinical Oncology, DOI 10.1007/978-1-60761-644-3_2, © Springer Science+Business Media, LLC 2011

17

18

G. Maschmeyer and P. Ljungman

Fig. 2.1  Infections following allogeneic hematopoietic stem cell transplantation (from Up to Date v18.3, Anaissie E, Marr KA, Thorner AR, 2010)

drug-related adverse events, allergy, and acute GvHD, must be considered. Infections in neutropenia after aHSCT may be life-­ threatening. Bacterial pathogens account for about 90% of infections during this phase. Epidemiological factors ­relevant for bacterial infections are shown in Table 2.1. Bacteremia, often related to central venous catheters (CVCs) and/or severe mucositis, occurs in up to 30% of patients after aHSCT, with the majority being caused by Gram-positive pathogens, predominantly coagulase-negative staphylococci, corynebacteria, and alpha-hemolytic streptococci [10–14]. Rarely, viridans streptococcal bacteremia may cause toxic shock and acute respiratory distress, potentially resulting in fatal outcome. Gram-negative infections are less frequent, but typically associated with higher morbidity and mortality. Gram-negative pathogens may enter the bloodstream via mucosal damage in the gastrointestinal tract of patients. Beyond that, fungal infection may occur in up to 15% of patients [15], and herpes simplex virus (HSV) infections emerge in this early posttransplant period unless acyclovir prophylaxis is given.

Table  2.1  Epidemiological aspects of bacterial infections after hematopoietic stem cell transplantation (HSCT) • Similar spectrum as neutropenic patients after intensive chemotherapy • Lower risk of severe and fatal infections early post-TxP after nonmyeloablative conditioning • Short neutropenia ± mucositis after autologous HSCT: infections comparable to other patients with short-term neutropenia, but Grampositive pathogens more frequent • Allogeneic HSCT: critical role of immune reconstitution −− Slow after T-cell depletion −− Slow after mismatched donor −− Slow/absent with significant graft-versus-host disease −− Chronic GvHD: functional asplenia

Diagnostic Procedures • Afebrile patient. −− Daily clinical exam + body temperature at least three times daily. Note:  antipyretic medication (steroids; analgesics such as metamizole).

2  Infections in Hematopoietic Stem Cell Transplant Recipients





















−− Serum C-reactive protein (CRP) twice weekly. −− Aspergillus antigen (GM) ³twice weekly. First fever. −− Update physical exam, blood cultures, clinical chemistry, CRP, interleukin-6 (IL-6), and thoracic computed tomography (CT) scan; other measures according to clinical findings (see below). Persistent fever. −− Update physical exam, blood cultures, clinical chemistry, CRP, IL-6, and thoracic CT scan; consider abdominal ultrasound or magnetic resonance imaging (MRI). −− Check results of antigen testings. Fever + pulmonary infiltrates. −− Bronchoscopy + bronchoalveolar lavage (BAL) =>microscopy + culture for bacteria; test for Mycobacterium tuberculous (MTB), Pneumocystis, cytomegalovirus (CMV), respiratory viruses, adenovirus, Aspergillus + other fungi; check for Aspergillus GM; optional: Aspergillus-PCR and MTB-/Pneumocystis-PCR. Fever + signs of inflammation at CVC. −− Blood cultures from peripheral vein and from CVC. −− Follow-up cultures in case of cultures positive for Staphylococcus aureus and Candida spp. Fever accompanied by skin lesions. −− Blood cultures. −− Biopsy (=>histopathology and nonfixated =>microbiology). Neurological symptoms ± fever. −− Cerebrospinal fluid (CSF) =>human herpes virus-6 (HHV-6); Aspergillus GM; CMV; HSV, VZV. −− Fundoscopy. −− Cranial MRI. Fever + abdominal symptoms. −− Clostridium difficile toxins; noro-/rotaviruses; CMV; adenovirus; Epstein–Barr virus (EBV). Perianal infiltrate/abscess. −− Beware of results from inappropriate microbiological diagnostics suggesting monomicrobial etiology. Fever + increasing “liver function tests” =>viral (hepatitis B virus (HBV), varicella zoster virus (VZV); CMV, etc.), Candida? −− Liver ultrasound or CT or MRI (preferred) [16]. NB:  Pneumocystis jiroveci typically accompanied by lactate dehydrogenase rise

If causative microorganisms have been isolated from blood, urine, or CSF culture, follow-up cultures should be obtained to document microbiological eradication, whenever possible. Since conventional chest radiography is insensitive and has a low negative predictive value for detecting pulmonary infiltrates in neutropenic patients, multislice or high-resolution CT of the lungs should be obtained early in neutropenic patients and particularly in those not responding to initial

19 Table  2.2  Pulmonary infections and noninfectious complications following allogeneic HSCT Early 90 days) Infectious (pneumonia) Noninfectious

Bacterial, fungal, viral, protozoal pathogens Pulmonary edema Idiopathic pneumonia syndrome Diffuse alveolar hemorrhage Engraftment syndrome Delayed pulmonary toxicity syndrome Secondary pulmonary alveolar proteinosis, pulmonary veno-occlusive disease

Bacterial, fungal, viral pathogens Restrictive lung disease Constrictive bronchiolitis Lymphocytic interstitial pneumonitis

antimicrobial therapy [17]. Differential diagnoses to pulmonary infiltrates posttransplant are shown in Table 2.2.

Antimicrobial Therapy in Patients with Neutropenic Fever After Allogeneic Stem Cell Transplantation Fever of more than 38.2°C, or fever of 38.0°C lasting for an hour or longer, or that recurs within 24 h should give reason for immediate broad-spectrum antibacterial treatment. Microbiological identification of an underlying pathogen is achievable in about one third of all patients. Therefore, it has become an accepted clinical practice to initiate broad-spectrum antimicrobial treatment empirically, or preemptively in the presence of specific clinical or radiological signs or symptoms. For selection of empiric antibacterial therapy in patients with febrile neutropenia, local antimicrobial resistance pattern must be taken into account. Initial empirical regimens should be active against enterobacteriaceae, Pseudomonas aeruginosa, S. aureus, and streptococci. Clinical trials that investigated single-agent regimens in patients with neutropenic fever included only few patients after allogeneic stem cell transplantation. Patients with severe mucositis should not be given single-agent ceftazidime because of the risk of bacteremia due to viridans streptococci, whereas piperacillin-tazobactam, imipenem, or meropenem appear appropriate. In the case of skin infections or venous catheter infections, prompt addition of a glycopeptide antibiotic to the initial empiric regimen should be considered. Stopping the administration of glycopeptides should be considered, if no multiresistant Gram-positive bacteria have been identified. In febrile neutropenic patients with pulmonary infiltrates, prompt preemptive addition of a systemic antifungal active against Aspergillus spp. is recommended [16].

20

Second-Line Empiric Antimicrobial Regimens in Patients with Neutropenic Fever After Allogeneic Stem Cell Transplantation If a causative infectious agent has been identified, modification of the empirically started antibacterial therapy according to the in vitro susceptibility pattern should be considered. In case of clinical nonresponse after 72–96 h of full-dose antibacterial treatment, modification of the antimicrobial regimen must be discussed and diagnostic procedures be repeated. Particularly in patients given a prednisone equivalent at a dose of >2  mg/kg/day, broad-spectrum systemic antifungal treatment should be part of the second-line treatment.

G. Maschmeyer and P. Ljungman Table  2.3  Risk factors of invasive fungal infection in patients after allogeneic SCT Early fungal infection (45 years Intensive immunosuppression as part of the conditioning regimen • Immunosuppression as prophylaxis and/or treatment of GvHD Late fungal infection (>40 days after SCT) • Immunosuppression due to GvHD and its treatment (corticosteroid or other more intensive immunosuppressive treatments) • Transplants from unrelated donors or family donors mismatched for HLA class I and/or class II antigens • Cytomegalovirus infections and antiviral therapy • Age >45 years

Duration of Antimicrobial Treatment Antimicrobial treatment may be discontinued if all of the following conditions are met: defervescence for at least 48 h, negative cultures, no clinical or radiological evidence of an infection, and neutrophil recovery to above 1,000/mL. If infections have been microbiologically proven, it is advisable to repeat the initial diagnostic procedures, in order to document the microbiological response (e.g., blood cultures, CSF cultures, urine cultures, stool cultures, bronchial secretions in case of ventilated patients, smears). In some cases, narrowing the antimicrobial spectrum can be acceptable.

Early Fungal Infections After Allogeneic Stem Cell Transplantation Epidemiological aspects of invasive fungal infections in transplant patients are listed in Table 2.3. In this patient population, the incidence rate of systemic mycoses can be as high as 15%, or higher under certain circumstances [15]. Increased risk is expected in patients with a previous history of invasive fungal infection, long-lasting severe neutropenia, previous episodes of prolonged neutropenia, severe skin and mucosal damages due to conditioning treatment, transplantation outside of a laminar air flow unit, age >45 years, intensive immunosuppression as part of the conditioning regimen or for prophylaxis, and/or treatment of GvHD [18]. Apart from specific local epidemiological conditions, Candida and Aspergillus species are predominant pathogens. Fever unresponsive to broad-spectrum antibiotic treatment may be the only early symptom of a systemic fungal infection. In patients with pulmonary Aspergillus infection, pleuritic chest pain, cough, or hemoptysis may occur. Blood cultures may occasionally grow Candida species. Aspergillus

spp. detected in clinical specimens (such as saliva or throat swabs) from neutropenic patients are likely to indicate incipient invasive infection. At the same time, even if moulds have been isolated from BAL specimens, it may be difficult to distinguish between contamination and true invasive pulmonary infection, whereas in cases of documented invasive pulmonary aspergillosis, cultures from BAL are often negative. Serial screening of blood samples for Aspergillus galactomannan or beta-d-glucan as well as for fungal DNA by polymerase chain reaction (PCR) may be helpful for early initiation of broad-spectrum systemic antifungal treatment. Antifungal agents frequently used in this situation are liposomal amphotericin B and caspofungin, both being licensed for empirical treatment of refractory neutropenic fever. If, however, thoracic CT scan shows typical findings indicative of invasive aspergillosis, voriconazole might be the first choice, as in case of probable or proven aspergillosis. Antifungal treatment is continued at least until neutrophil recovery and resolution of clinical and radiological signs of infection. Other mould infections such as zygomycosis and fusariosis are rare, but increasingly reported in patients post-aHSCT, and in case of suspected zygomycosis, liposomal amphotericin B would be the preferred choice.

Early Viral Infections After Allogeneic Stem Cell Transplantation Virus infections can occur during the period before hematopoietic engraftment. HSV reactivates frequently in this early period unless acyclovir prophylaxis is given, and the clinical symptoms are frequently uncharacteristic [19]. Acyclovir-resistant viruses have been reported in different

21

2  Infections in Hematopoietic Stem Cell Transplant Recipients

patient series to occur in up to 10% and should be suspected if mucositis is prolonged in patients on acyclovir prophylaxis [20, 21]. Respiratory viral infections especially caused by RSV, parainfluenza, and influenza can occur early and are frequently due to nosocomial transmission within the transplant unit and therefore infection control procedures should be in place during times due to community outbreaks of these viruses [22–27]. Lower respiratory tract infection due to RSV and parainfluenza are associated with significant mortality. Patients, who are HBV DNA positive or HBsAg positive, before aHSCT are at risk for severe hepatitis and should be given prolonged antiviral prophylaxis [28–30].

Intermediate Posttransplant Period (3 Weeks to 3 Months) Specific Epidemiology of Infections in the Intermediate Posttransplant Period In the majority of allogeneic stem cell transplant recipients, infections emerge later than day +50 posttransplant. After hematopoietic reconstitution, a severe combined quantitative and functional deficiency in the T and B lymphocyte ­compartment persists. If T-cell depletion has been used, or if HLA-incompatibility between recipient and donor had to be accepted, immunodeficiency will be prolonged after ­transplantation. Immunodeficiency comprises impaired T helper cell function, immunoglobulin synthesis, and ­cytotoxic T cell response. Despite normalization of white blood cell counts, compromised granulocyte functions, primarily impairment of chemotaxis and phagocytosis, may persist.

Bacterial and Fungal Infections in the Intermediate Posttransplantation Period In 14% of patients, bacteremia occurs after hematopoietic engraftment, with a mortality rate comparable to that before and after engraftment. Among blood culture isolates, Grampositive pathogens (staphylococci in particular) are predominant, with the focus being identified in more than 50% of patients. Venous catheter infections are the cause for more than 30% of bacteremias, and fever and chills within the first hour after start of fluid infusion typically are indicating a catheter-related bacteremia. Other more frequent infections during the intermediate posttransplant period are pneumonias, preferably caused by Streptococcus pneumoniae, Klebsiella species, and P. aeruginosa, or by filamentous fungi such as Aspergillus. Among less frequent bacterial

pathogens relevant during this period are Listeria monocytogenes and Legionella pneumophila. While listeriosis may origin from products made from unpasteurized milk, the latter typically is related to the use of showers or jacuzzis after the water has been resting in the pipes for a longer period of time. In patients who are treated with tumor necrosis factor antagonists such as infliximab, a dramatic increase in the risk of invasive fungal infections must be considered [31, 32]. Apart from aspergillosis, some rare forms of invasive mycosis caused by Fusarium spp., zygomycetes, resistant Candida spp., Pseudallescheria boydii (or its asexual form, Scedosporium apiospermum), and others may occur during this time period [33]. Typically, patients with fusariosis have skin lesions and positive blood cultures, while zygomycetes cause clinical syndromes resembling aspergillosis.

Viral Infections Viruses are common causes of infections during the period from engraftment to day +90 after HSCT. The classic viral pathogen during this period is CMV called “the troll of transplantation.” CMV reactivates in 60–70% of pretransplant seropositive patients and primary infections occur in up to one third of seronegative patients with seropositive donors [34]. Established end-organ CMV disease is still associated with significant morbidity and mortality. Therefore, preventive strategies either by antiviral prophylaxis or preemptive therapy should be used [35, 36]. Antiviral prophylaxis has been less used, but new antiviral agents might make this strategy more attractive. Monitoring with sensitive assays such as pp65 antigenemia or quantitative PCR in blood is indicated in all aHSCT recipients to allow early initiation of antiviral therapy with ganciclovir or valganciclovir [35]. Epstein-Barr Virus (EBV) also reactivates very frequently after aHSCT, but rarely causes end-organ disease [37]. However, EBV-driven posttransplant lymphoproliferative disease (PTLD) is a complication with high mortality unless treated [38–41]. This complication is more commonly seen in EBV seronegative patients receiving grafts from EBV seropositive donors and in patients having delayed immune reconstitution such as after a T-cell-depleted or HLA-mismatched stem cell transplantation. PTLD frequently causes unspecific symptoms frequently with fever and lymphadenopathy and is associated with high levels of EBV in blood [39, 40, 42–44]. Rituximab (anti CD20 antibody) given either as preemptive therapy or as therapy for established PTLD is most likely effective, although no controlled trial has been performed [45–49]. Adenovirus infections can cause multiorgan disease including pneumonia, encephalitis, hepatitis, gastroenteritis, and hemorrhagic cystitis. Severe adenovirus disease is more ­frequently seen in children especially after transplant

22

procedures­resulting in delayed reconstitution of the immune ­system such as haploidentical transplants or mismatched cord blood grafts and monitoring for adenovirus in blood might be indicated in such patients [50–57]. Cidofovir is a potentially effective antiviral agent, but is associated with nephrotoxicity [58–60]. Other viral pathogens potentially important during this period after HSCT are HHV-6 associated with encephalitis and bone marrow suppression [61–66], BK-virus infections associated with hemorrhagic cystitis [67, 68], and respiratory viruses.

Late Period After Allogeneic Stem Cell Transplantation (Later than Day +90) Specific Epidemiology of Infections During the Late Posttransplant Period In the late posttransplant period, immune reconstitution usually advances, particularly in patients who have received a transplant from an HLA-identical family donor. These patients often show full hematopoietic reconstitution and early immune reconstitution. If no relevant GVHD emerges, prophylactic immunosuppression will typically be discontinued. Patients with a CD4-count of >200/mL blood and normalized serum immunoglobulin levels can be considered as immunocompetent without an increased risk of opportunistic infections. However, in the case of chronic GvHD, which may occur in more than 30% of patients, a severe combined cellular and humoral immunodeficiency will persist for a prolonged period of time. Mucosal damage, functional deficiencies of granulocytes (especially impaired chemotaxis), functional asplenia, and qualitative as well as quantitative T- and B-cell deficiencies pave the way to a significantly increased susceptibility to infections in these patients. In particular, bacterial infections of the respiratory tract constitute a major cause of death [69]. Life-threatening infections are typically caused by encapsulated bacteria such as S. pneumoniae or Haemophilus influenzae. Sinusitis, otitis media, and pharyngitis may indicate such infections in the late posttransplant period. Patients among this risk group presenting with signs of infection should receive immediate antibacterial treatment. An important pathogen of interstitial pneumonia in the late phase after allogeneic stem cell transplantation is P. jiroveci [70]. Without specific prophylaxis, about 30% of patients with chronic GvHD develop Pneumocystis pneumonia, which can take a fatal course in up to 15% of patients and prophylaxis given for at least 6 months (and longer in patients with chronic GvHD) is recommended to all patients. In regions

G. Maschmeyer and P. Ljungman

with relatively high prevalence rates, mycobacterial ­infections should be taken into consideration as well [71, 72].

Late Viral Infections After Stem Cell Transplantation Late-occurring CMV infection and disease have become more frequent during the last decade. These are associated with delayed and incomplete reconstitution of specific immunity, primarily of T-cells, and occur more commonly in patients experiencing severe GvHD [73, 74]. Prolonged monitoring and repeated antiviral therapy are needed in such patients, although toxicity from antiviral therapy and development of resistant CMV strains are important considerations [34, 36]. The possibility to reconstitute specific immunity by adoptive transfer of T-cells has been explored by several groups. VZV is an important pathogen after HSCT. Primary varicella – chickenpox – occurring in seronegative patients is an important complication especially in children. Preventive measures should be taken after exposure and i.v. acyclovir therapy given if the infection develops. VZV can reactivate also early after aHSCT, but infections are more commonly seen during the late posttransplant period. The clinical manifestations vary from localized herpes zoster – shingles – to visceral disseminated disease associated with high mortality [75–77]. Visceral disease including CNS disease can occur without cutaneous manifestations and can therefore be difficult to diagnose. Early initiation of antiviral therapy with intravenous acyclovir is ­crucial when visceral or disseminated VZV disease is suspected. Localized shingles can often be treated with orally given valacyclovir or famciclovir [37]. In many centers, longterm prophylaxis given for at least one year after HSCT is used to prevent VZV reactivations [78, 79]. Respiratory viruses, especially influenza, can also be severe late after HSCT. Yearly vaccination against influenza is therefore recommended [80]. RSV and parainfluenza infections have been associated with late respiratory compromise presumably through immune-mediated mechanisms [81, 82]. HBV infection can reactivate in previously HBV-infected patients, especially during prolonged treatment for GvHD. Reactivation can result in a potentially severe acute hepatitis and patients should be carefully monitored, and if signs of HBV reactivation develop, be given antiviral therapy [28–30].

Late Fungal Infections After Stem Cell Transplant Fungal infections during late transplant period are discussed in detail in Part III.

2  Infections in Hematopoietic Stem Cell Transplant Recipients

References 1. Walter E, Bowden RA. Infection in the bone marrow transplant recipient. Infect Dis Clin North Am. 1995;9:823–47. 2. Krüger WH, Bohlius J, Cornely OA, et al. Antimicrobial prophylaxis in allogeneic bone marrow transplantation. Guidelines of the infectious diseases working party (AGIHO) of the German Society of Haematology and Oncology. Ann Oncol. 2005;16:1381–90. 3. Tomblyn M, Chiller T, Einsele H, et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant. 2009;15:1143–238. 4. Welniak LA, Blazar BR, Murphy WJ. Immunobiology of allogeneic­ hematopoietic stem cell transplantation. Annu Rev Immunol. 2007;25:139–70. 5. Aschan J. Risk assessment in haematopoietic stem cell transplantation: conditioning. Best Pract Res Clin Haematol. 2007;20:295–310. 6. Bachanova V, Brunstein CG, Burns LJ, et al. Fewer infections and lower infection-related mortality following non-myeloablative versus myeloablative conditioning for allotransplantation of patients with lymphoma. Bone Marrow Transplant. 2009;43:237–44. 7. Baron F, Storb R. Hematopoietic cell transplantation after reducedintensity conditioning for older adults with acute myeloid leukemia in complete remission. Curr Opin Hematol. 2007;14:145–51. 8. Junghanss C, Marr KA, Carter RA, et al. Incidence and outcome of bacterial and fungal infections following nonmyeloablative compared with myeloablative allogeneic hematopoietic stem cell transplantation: a matched control study. Biol Blood Marrow Transplant. 2002;8:512–20. 9. Meijer E, Dekker AW, Lokhorst HM, Petersen EJ, Nieuwenhuis HK, Verdonck LF. Low incidence of infectious complications after nonmyeloablative compared with myeloablative allogeneic stem cell transplantation. Transpl Infect Dis. 2004;6:171–8. 10. Blijlevens NM, Donnelly JP, de Pauw BE. Prospective evaluation of gut mucosal barrier injury following various myeloablative regimens for haematopoietic stem cell transplant. Bone Marrow Transplant. 2005;35:707–11. 11. Collin BA, Leather HL, Wingard JR, Ramphal R. Evolution, incidence, and susceptibility of bacterial bloodstream isolates from 519 bone marrow transplant patients. Clin Infect Dis. 2001;33:947–53. 12. Kolbe K, Domkin D, Derigs HG, Bhakdi S, Huber C, Aulitzky WE. Infectious complications during neutropenia subsequent to peripheral blood stem cell transplantation. Bone Marrow Transplant. 1997;19:143–7. 13. Krüger W, Rüssmann B, Kröger N, et al. Early infections in patients undergoing bone marrow or blood stem cell transplantation – a 7 year single centre investigation of 409 cases. Bone Marrow Transplant. 1999;23:589–97. 14. Yuen KY, Woo PCY, Hui CH, et al. Unique risk factors for bacteraemia in allogeneic bone marrow transplant recipients before and after engraftment. Bone Marrow Transplant. 1998;21:1137–43. 15. Marr KA, Carter RA, Boeckh M, Martin P, Corey L. Invasive aspergillosis in allogeneic stem cell transplant recipients: changes in epidemiology and risk factors. Blood. 2002;100:4358–66. 16. Einsele H, Bertz H, Beyer J, et  al. Infectious complications after allogeneic stem cell transplantation - Epidemiology and interventional therapy strategies. Ann Hematol. 2003;82 Suppl 2:S175–85. 17. Heussel CP, Kauczor HU, Heussel GE, et al. Pneumonia in febrile neutropenic patients and in bone marrow and blood stem-cell transplant recipients: use of high-resolution computed tomography. J Clin Oncol. 1999;17:796–805. 18. Barnes PD, Marr KA. Risks, diagnosis and outcomes of invasive fungal infections in haematopoietic stem cell transplant recipients. Br J Haematol. 2007;139:519–31.

23 19. Wade JC, Day LM, Crowley JJ, Meyers JD. Recurrent infection with herpes simplex virus after marrow transplantation: role of the specific immune response and acyclovir treatment. J Infect Dis. 1984;149:750–6. 20. Chakrabarti S, Pillay D, Ratcliffe D, Cane PA, Collingham KE, Milligan DW. Resistance to antiviral drugs in herpes simplex virus infections among allogeneic stem cell transplant recipients: risk factors and prognostic significance. J Infect Dis. 2000; 181:2055–8. 21. Chen Y, Scieux C, Garrait V, et al. Resistant herpes simplex virus type 1 infection: an emerging concern after allogeneic stem cell transplantation. Clin Infect Dis. 2000;31:927–35. 22. Harrington RD, Hooton TM, Hackman RC, et al. An outbreak of respiratory syncytial virus in a bone marrow transplant center. J Infect Dis. 1992;165:987–93. 23. Ljungman P, Ward KN, Crooks BN, et al. Respiratory virus infections after stem cell transplantation: a prospective study from the infectious diseases working party of the European group for blood and marrow transplantation. Bone Marrow Transplant. 2001;28:479–84. 24. McCann S, Byrne JL, Rovira M, et  al. Outbreaks of infectious diseases in stem cell transplant units: a silent cause of death for patients and transplant programmes. Bone Marrow Transplant. 2004;33:519–29. 25. Nichols WG, Corey L, Gooley T, Davis C, Boeckh M. Parainfluenza virus infections after hematopoietic stem cell transplantation: risk factors, response to antiviral therapy, and effect on transplant outcome. Blood. 2001;98:573–8. 26. Nichols WG, Guthrie KA, Corey L, Boeckh M. Influenza infections after hematopoietic stem cell transplantation: risk factors, mortality, and the effect of antiviral therapy. Clin Infect Dis. 2004; 39:1300–6. 27. Machado CM. Influenza infections after hematopoietic stem cell transplantation. Clin Infect Dis. 2005;41:273–4. 28. Locasciulli A, Bruno B, Alessandrino EP, et al. Hepatitis reactivation and liver failure in haemopoietic stem cell transplants for hepatitis B virus (HBV)/hepatitis C virus (HCV) positive recipients: a retrospective study by the Italian group for blood and marrow transplantation. Bone Marrow Transplant. 2003;31:295–300. 29. Kitano K, Kobayashi H, Hanamura M, et  al. Fulminant hepatitis after allogenic bone marrow transplantation caused by reactivation of hepatitis B virus with gene mutations in the core promotor region. Eur J Haematol. 2006;77:255–8. 30. Hsiao LT, Chiou TJ, Liu JH, et  al. Extended lamivudine therapy against hepatitis B virus infection in hematopoietic stem cell transplant recipients. Biol Blood Marrow Transplant. 2006;12:84–94. 31. Hamadani M, Hofmeister CC, Jansak B, et al. Addition of infliximab to standard acute graft-versus-host disease prophylaxis following allogeneic peripheral blood cell transplantation. Biol Blood Marrow Transplant. 2008;14:783–9. 32. Marty FM, Lee SJ, Fahey MM, et al. Infliximab use in patients with severe graft-versus-host disease and other emerging risk factors of nonCandida invasive fungal infections in allogeneic hematopoietic stem cell transplant recipients: a cohort study. Blood. 2003;102:2768–76. 33. Marr KA, Carter RA, Crippa F, Wald A, Corey L. Epidemiology and outcome of mould infections in hematopoietic stem cell transplant recipients. Clin Infect Dis. 2002;34:909–17. 34. Boeckh M, Ljungman P. How we treat cytomegalovirus in hematopoietic cell transplant recipients. Blood. 2009;113:5711–9. 35. Ljungman P, de la Camara R, Cordonnier C, et al. Management of CMV, HHV-6, HHV-7 and Kaposi-sarcoma herpesvirus (HHV-8) infections in patients with hematological malignancies and after SCT. Bone Marrow Transplant. 2008;42:227–40. 36. Zaia J, Baden L, Boeckh MJ, et al. Viral disease prevention after hematopoietic cell transplantation. Bone Marrow Transplant. 2009; 44:471–82.

24 37. Styczynski J, Reusser P, Einsele H, et  al. Management of HSV, VZV and EBV infections in patients with hematological malignancies and after SCT: guidelines from the second European conference on infections in Leukemia. Bone Marrow Transplant. 2009; 43:757–70. 38. Micallef IN, Chanabhai M, Gascoyne RD, et al. Lymphoproliferative disorders following allogeneic bone marrow transplantation: the Vancouver experience. Bone Marrow Transplant. 1998;22:981–7. 39. Juvonen E, Aalto SM, Tarkkanen J, et al. High incidence of PTLD after non-T-cell-depleted allogeneic haematopoietic stem cell transplantation as a consequence of intensive immunosuppressive treatment. Bone Marrow Transplant. 2003;32:97–102. 40. Sundin M, Le Blanc K, Ringden O, et  al. The role of HLA mismatch, splenectomy and recipient Epstein–Barr virus seronegativity as risk factors in post-transplant lymphoproliferative disorder following allogeneic hematopoietic stem cell transplantation. Haematologica. 2006;91:1059–67. 41. Landgren O, Gilbert ES, Rizzo JD, et al. Risk factors for lymphoproliferative disorders after allogeneic hematopoietic cell transplantation. Blood. 2009;113:4992–5001. 42. Gartner BC, Schafer H, Marggraff K, et  al. Evaluation of use of Epstein–Barr viral load in patients after allogeneic stem cell transplantation to diagnose and monitor posttransplant lymphoproliferative disease. J Clin Microbiol. 2002;40:351–8. 43. Greenfield HM, Gharib MI, Turner AJ, et al. The impact of monitoring Epstein–Barr virus PCR in paediatric bone marrow transplant patients: can it successfully predict outcome and guide intervention? Pediatr Blood Cancer. 2006;47:200–5. 44. Kinch A, Oberg G, Arvidson J, Falk KI, Linde A, Pauksens K. Posttransplant lymphoproliferative disease and other Epstein–Barr virus diseases in allogeneic haematopoietic stem cell transplantation after introduction of monitoring of viral load by polymerase chain reaction. Scand J Infect Dis. 2007;39:235–44. 45. Van Esser JW, Niesters HG, van der Holt B, et  al. Prevention of Epstein–Barr virus-lymphoproliferative disease by molecular monitoring and preemptive rituximab in high-risk patients after allogeneic stem cell transplantation. Blood. 2002;99:4364–9. 46. Wagner HJ, Cheng YC, Huls MH, et al. Prompt versus preemptive intervention for EBV lymphoproliferative disease. Blood. 2004;103:3979–81. 47. Weinstock DM, Ambrossi GG, Brennan C, Kiehn TE, Jakubowski A. Preemptive diagnosis and treatment of Epstein–Barr virus-associated post transplant lymphoproliferative disorder after hematopoietic stem cell transplant: an approach in development. Bone Marrow Transplant. 2006;37:539–46. 48. Styczynski J, Einsele H, Gil L, Ljungman P. Outcome of treatment of Epstein–Barr virus-related post-transplant lymphoproliferative disorder in hematopoietic stem cell recipients: a comprehensive review of reported cases. Transpl Infect Dis. 2009;11:383–92. 49. Omar H, Hagglund H, Gustafsson-Jernberg A, et al. Targeted monitoring of patients at high risk of post-transplant lymphoproliferative disease by quantitative Epstein–Barr virus polymerase chain reaction. Transpl Infect Dis. 2009;11:393–9. 50. Shields AF, Hackman RC, Fife KH, Corey L, Meyers JD. Adenovirus infections in patients undergoing bone-marrow transplantation. N Engl J Med. 1985;312:529–33. 51. Flomenberg P, Babbitt J, Drobyski WR, et al. Increasing incidence of adenovirus disease in bone marrow transplant recipients. J Infect Dis. 1994;169:775–81. 52. Chakrabarti S, Mautner V, Osman H, et al. Adenovirus infections following allogeneic stem cell transplantation: incidence and outcome in relation to graft manipulation, immunosuppression, and immune recovery. Blood. 2002;100:1619–27. 53. Lion T, Baumgartinger R, Watzinger F, et al. Molecular monitoring of adenovirus in peripheral blood after allogeneic bone marrow

G. Maschmeyer and P. Ljungman transplantation permits early diagnosis of disseminated disease. Blood. 2003;102:1114–20. 54. Van Tol MJ, Kroes AC, Schinkel J, et al. Adenovirus infection in paediatric stem cell transplant recipients: increased risk in young children with a delayed immune recovery. Bone Marrow Transplant. 2005;36:39–50. 55. Feuchtinger T, Lang P, Handgretinger R. Adenovirus infection after allogeneic stem cell transplantation. Leuk Lymphoma. 2007;48:244–55. 56. Robin M, Marque-Juillet S, Scieux C, et al. Disseminated adenovirus infections after allogeneic hematopoietic stem cell transplantation: incidence, risk factors and outcome. Haematologica. 2007;92:1254–7. 57. Symeonidis N, Jakubowski A, Pierre-Louis S, Jaffe D, Pamer E, Sepkowitz K, et al. Invasive adenoviral infections in T-cell-depleted allogeneic hematopoietic stem cell transplantation: high mortality in the era of cidofovir. Transpl Infect Dis. 2007;9:108–13. 58. Ljungman P, Ribaud P, Eyrich M, et  al. Cidofovir for adenovirus infections after allogeneic hematopoietic stem cell transplantation: a survey by the infectious diseases working party of the European group for blood and marrow transplantation. Bone Marrow Transplant. 2003;31:481–6. 59. Yusuf U, Hale GA, Carr J, et al. Cidofovir for the treatment of adenoviral infection in pediatric hematopoietic stem cell transplant patients. Transplantation. 2006;81:1398–404. 60. Neofytos D, Ojha A, Mookerjee B, et al. Treatment of adenovirus disease in stem cell transplant recipients with cidofovir. Biol Blood Marrow Transplant. 2007;13:74–81. 61. Drobyski WR, Dunne WM, Burd EM, et al. Human herpesvirus-6 (HHV-6) infection in allogeneic bone marrow transplant recipients: evidence of a marrow-suppressive role for HHV-6 in vivo. J Infect Dis. 1993;167:735–9. 62. Carrigan DR, Knox KK. Human herpesvirus 6 (HHV-6) isolation from bone marrow: HHV-6-associated bone marrow suppression in bone marrow transplant patients. Blood. 1994;84:3307–10. 63. Wang FZ, Linde A, Hagglund H, Testa M, Locasciulli A, Ljungman P. Human herpesvirus 6 DNA in cerebrospinal fluid specimens from allogeneic bone marrow transplant patients: does it have clinical significance? Clin Infect Dis. 1999;28:562–8. 64. Zerr DM, Gooley TA, Yeung L, et al. Human herpesvirus 6 reactivation and encephalitis in allogeneic bone marrow transplant recipients. Clin Infect Dis. 2001;33:763–71. 65. Zerr DM, Gupta D, Huang ML, Carter R, Corey L. Effect of antivirals on human herpesvirus 6 replication in hematopoietic stem cell transplant recipients. Clin Infect Dis. 2002;34:309–17. 66. Zerr DM, Corey L, Kim HW, Huang ML, Nguy L, Boeckh M. Clinical outcomes of human herpesvirus 6 reactivation after hematopoietic stem cell transplantation. Clin Infect Dis. 2005;40:932–40. 67. Leung AY, Suen CK, Lie AK, Liang RH, Yuen KY, Kwong YL. Quantification of polyoma BK viruria in hemorrhagic cystitis complicating bone marrow transplantation. Blood. 2001;98:1971–8. 68. Erard V, Kim HW, Corey L, et al. BK DNA viral load in plasma: evidence for an association with hemorrhagic cystitis in allogeneic hematopoietic cell transplant recipients. Blood. 2005;106:1130–2. 69. Chen CS, Boeckh M, Seidel K, et al. Incidence, risk factors, and mortality from pneumonia developing late after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2003;32:515–22. 70. De Castro N, Neuville S, Sarfati C, et al. Occurrence of Pneumocystis jiroveci pneumonia after allogeneic stem cell transplantation: a 6-year retrospective study. Bone Marrow Transplant. 2005;36:879–83. 71. Cordonnier C, Martino R, Trabasso P, et al. Mycobacterial infection: a difficult and late diagnosis in stem cell transplant recipients. Clin Infect Dis. 2004;38:1229–36. 72. Erdstein AA, Daas P, Bradstock KF, Robinson T, Hertzberg MS. Tuberculous in allogeneic stem cell transplant recipients: still a problem in the 21st century. Transpl Infect Dis. 2004;6:142–6.

2  Infections in Hematopoietic Stem Cell Transplant Recipients 73. Boeckh M, Leisenring W, Riddell SR, et al. Late cytomegalovirus disease and mortality in recipients of allogeneic hematopoietic stem cell transplants: importance of viral load and T-cell immunity. Blood. 2003;101:407–14. 74. Hakki M, Riddell SR, Storek J, et al. Immune reconstitution to cytomegalovirus after allogeneic hematopoietic stem cell transplantation: impact of host factors, drug therapy, and subclinical reactivation. Blood. 2003;102:3060–7. 75. Locksley RM, Flournoy N, Sullivan KM, Meyers JD. Infection with varicella-zoster virus after marrow transplantation. J Infect Dis. 1985;152:1172–81. 76. Steer CB, Szer J, Sasadeusz J, Matthews JP, Beresford JA, Grigg A. Varicella-zoster infection after allogeneic bone marrow transplantation: incidence, risk factors and prevention with low-dose aciclovir and ganciclovir. Bone Marrow Transplant. 2000;25:657–64. 77. Martino R, Rovira M, Carreras E, et al. Severe infections after allogeneic peripheral blood stem cell transplantation: a matched-pair comparison of unmanipulated and CD34+ cell-selected transplantation. Haematologica. 2001;86:1075–86.

25 78. Boeckh M, Kim HW, Flowers ME, Meyers JD, Bowden RA. Long-term acyclovir for prevention of varicella zoster virus ­disease after allogeneic hematopoietic cell transplantation – a ­randomized double-blind placebo-controlled study. Blood. 2006;107:1800–5. 79. Erard V, Guthrie KA, Varley C, et  al. One-year acyclovir ­prophylaxis for preventing varicella-zoster virus (VZV) disease following hematopoietic cell transplantation: no evidence of rebound VZV disease after drug discontinuation. Blood. 2007; 110:3071–7. 80. Ljungman P, Cordonnier C, Einsele H, Englund J, Machado CM, Storek J, et al. Vaccination of hematopoietic cell transplant recipients. Bone Marrow Transplant. 2009;44:521–6. 81. Erard V, Chien JW, Kim HW, et al. Airflow decline after myeloablative allogeneic hematopoietic cell transplantation: the role of community respiratory viruses. J Infect Dis. 2006;193:1619–25. 82. Avetisyan G, Mattsson J, Sparrelid E, Ljungman P. Respiratory syncytial virus infection in recipients of allogeneic stem-cell transplantation: a retrospective study of the incidence, clinical features, and outcome. Transplantation. 2009;88:1222–6.

http://www.springer.com/978-1-60761-643-6

Suggest Documents